1
|
Elia N, Prinelli F, Peli V, Conti S, Barilani M, Mei C, Castaldi S, Lazzari L. Public attitudes toward the use of human induced pluripotent stem cells: insights from an Italian adult population. Front Public Health 2024; 12:1491257. [PMID: 39568604 PMCID: PMC11576450 DOI: 10.3389/fpubh.2024.1491257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 10/25/2024] [Indexed: 11/22/2024] Open
Abstract
Introduction Human induced pluripotent stem cells (hiPSCs), derived from reprogrammed adult somatic cells, hold significant promise for disease modelling, personalized medicine, drug discovery, and regenerative therapies. Public awareness and understanding of hiPSCs are crucial for advancing research in this field. However, limited data exists on the general population's knowledge and attitudes toward their use. Methods This study aimed to assess the awareness and perceptions of hiPSCs among Italian adults through a web-based survey conducted via the EUSurvey platform, using a snowball sampling approach. The survey included demographic information and mandatory questions on knowledge, awareness, and concerns regarding hiPSC technology, with responses collected on a 3-point scale. Statistical analysis was performed using chi-squared tests, with significance set at p ≤ 0.05. Results Out of 1874 respondents, the majority were aged 18-35 years (40.5%), female (63.4%), and university-educated (67.2%). Among those familiar with hiPSCs (54.1%, n = 1,201), 95.3% expressed willingness to donate blood samples for hiPSC generation to treat individuals with incurable diseases. Concerns about current research and therapeutic applications were low (less than 20%), but nearly half of the respondents were hesitant or opposed to the use of hiPSCs in animal experiments and their commercialization by pharmaceutical companies. Increased skepticism was observed in older, less educated, religious individuals, and those who were not blood donors. Overall, the Italian public shows strong support for hiPSC-based therapies, though reservations exist around specific ethical and economic issues. Discussion These findings underscore the importance of addressing public concerns through targeted educational campaigns, not only in Italy but globally, to foster a more informed and supportive environment for advancing stem cell research and its clinical applications worldwide. Similar studies have been conducted in Japan, the United States, and Sweden, but there remains a need for all countries to engage with their citizens to better understand how stem cell research is perceived locally. Such engagement is crucial for guiding international strategies in personalized medicine and regenerative therapies, ensuring that emerging technologies are met with both ethical integrity and public trust.
Collapse
Affiliation(s)
- Noemi Elia
- Unit of Cell and Gene Therapy, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Federica Prinelli
- Epidemiology and Public Health Unit, Institute of Biomedical Technologies - National Research Council, Segrate (MI), Italy
| | - Valeria Peli
- Unit of Cell and Gene Therapy, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Silvia Conti
- Epidemiology and Public Health Unit, Institute of Biomedical Technologies - National Research Council, Segrate (MI), Italy
| | - Mario Barilani
- Unit of Cell and Gene Therapy, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Cecilia Mei
- Unit of Cell and Gene Therapy, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Dino Ferrari Center, Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Silvana Castaldi
- Quality Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Lorenza Lazzari
- Unit of Cell and Gene Therapy, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
2
|
Rehman A, Fatima I, Noor F, Qasim M, Wang P, Jia J, Alshabrmi FM, Liao M. Role of small molecules as drug candidates for reprogramming somatic cells into induced pluripotent stem cells: A comprehensive review. Comput Biol Med 2024; 177:108661. [PMID: 38810477 DOI: 10.1016/j.compbiomed.2024.108661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/08/2024] [Accepted: 05/26/2024] [Indexed: 05/31/2024]
Abstract
With the use of specific genetic factors and recent developments in cellular reprogramming, it is now possible to generate lineage-committed cells or induced pluripotent stem cells (iPSCs) from readily available and common somatic cell types. However, there are still significant doubts regarding the safety and effectiveness of the current genetic methods for reprogramming cells, as well as the conventional culture methods for maintaining stem cells. Small molecules that target specific epigenetic processes, signaling pathways, and other cellular processes can be used as a complementary approach to manipulate cell fate to achieve a desired objective. It has been discovered that a growing number of small molecules can support lineage differentiation, maintain stem cell self-renewal potential, and facilitate reprogramming by either increasing the efficiency of reprogramming or acting as a genetic reprogramming factor substitute. However, ongoing challenges include improving reprogramming efficiency, ensuring the safety of small molecules, and addressing issues with incomplete epigenetic resetting. Small molecule iPSCs have significant clinical applications in regenerative medicine and personalized therapies. This review emphasizes the versatility and potential safety benefits of small molecules in overcoming challenges associated with the iPSCs reprogramming process.
Collapse
Affiliation(s)
- Abdur Rehman
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, PR China
| | - Israr Fatima
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, PR China
| | - Fatima Noor
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan; Department of Bioinformatics and Biotechnology, Government College University of Faisalabad, 38000, Pakistan
| | - Muhammad Qasim
- Department of Bioinformatics and Biotechnology, Government College University of Faisalabad, 38000, Pakistan
| | - Peng Wang
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, PR China
| | - Jinrui Jia
- Laboratory of Animal Fat Deposition and Muscle Development, Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, PR China
| | - Fahad M Alshabrmi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, 51452, Saudi Arabia
| | - Mingzhi Liao
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, PR China.
| |
Collapse
|
3
|
Cell models for Down syndrome-Alzheimer’s disease research. Neuronal Signal 2022; 6:NS20210054. [PMID: 35449591 PMCID: PMC8996251 DOI: 10.1042/ns20210054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 03/07/2022] [Accepted: 03/21/2022] [Indexed: 11/29/2022] Open
Abstract
Down syndrome (DS) is the most common chromosomal abnormality and leads to intellectual disability, increased risk of cardiac defects, and an altered immune response. Individuals with DS have an extra full or partial copy of chromosome 21 (trisomy 21) and are more likely to develop early-onset Alzheimer’s disease (AD) than the general population. Changes in expression of human chromosome 21 (Hsa21)-encoded genes, such as amyloid precursor protein (APP), play an important role in the pathogenesis of AD in DS (DS-AD). However, the mechanisms of DS-AD remain poorly understood. To date, several mouse models with an extra copy of genes syntenic to Hsa21 have been developed to characterise DS-AD-related phenotypes. Nonetheless, due to genetic and physiological differences between mouse and human, mouse models cannot faithfully recapitulate all features of DS-AD. Cells differentiated from human-induced pluripotent stem cells (iPSCs), isolated from individuals with genetic diseases, can be used to model disease-related cellular and molecular pathologies, including DS. In this review, we will discuss the limitations of mouse models of DS and how these can be addressed using recent advancements in modelling DS using human iPSCs and iPSC-mouse chimeras, and potential applications of iPSCs in preclinical studies for DS-AD.
Collapse
|
4
|
Alsaloum M, Waxman SG. iPSCs and DRGs: stepping stones to new pain therapies. Trends Mol Med 2022; 28:110-122. [PMID: 34933815 PMCID: PMC8810720 DOI: 10.1016/j.molmed.2021.11.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 02/03/2023]
Abstract
There is a pressing need for more effective nonaddictive treatment options for pain. Pain signals are transmitted from the periphery into the spinal cord via dorsal root ganglion (DRG) neurons, whose excitability is driven by voltage-gated sodium (NaV) channels. Three NaV channels (NaV1.7, NaV1.8, and NaV1.9), preferentially expressed in DRG neurons, play important roles in pain signaling in humans. Blockade of these channels may provide a novel approach to the treatment of pain, but clinical translation of preclinical results has been challenging, in part due to differences between rodent and human DRG neurons. Human DRG neurons and iPSC-derived sensory neurons (iPSC-SNs) provide new preclinical platforms that may facilitate the development of novel pain therapeutics.
Collapse
Affiliation(s)
- Matthew Alsaloum
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA; Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA; Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA; Yale Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, USA; Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA; Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA; Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA.
| |
Collapse
|
5
|
Duckert B, Vinkx S, Braeken D, Fauvart M. Single-cell transfection technologies for cell therapies and gene editing. J Control Release 2020; 330:963-975. [PMID: 33160005 DOI: 10.1016/j.jconrel.2020.10.068] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/30/2020] [Accepted: 10/31/2020] [Indexed: 12/29/2022]
Abstract
Advances in gene editing and cell therapies have recently led to outstanding clinical successes. However, the lack of a cost-effective manufacturing process prevents the democratization of these innovative medical tools. Due to the common use of viral vectors, the step of transfection in which cells are engineered to gain new functions, is a major bottleneck in making safe and affordable cell products. A promising opportunity lies in Single-Cell Transfection Technologies (SCTTs). SCTTs have demonstrated higher efficiency, safety and scalability than conventional transfection methods. They can also feature unique abilities such as substantial dosage control over the cargo delivery, single-cell addressability and integration in microdevices comprising multiple monitoring modalities. Unfortunately, the potential of SCTTs is not fully appreciated: they are most often restricted to research settings with little adoption in clinical settings. To encourage their adoption, we review and compare recent developments in SCTTs, and how they can enable selected clinical applications. To help bridge the gap between fundamental research and its translation to the clinic, we also describe how Good Manufacturing Practices (GMP) can be integrated in the design of SCTTs.
Collapse
Affiliation(s)
- Bastien Duckert
- Department of Physics and Astronomy, KU Leuven, Celestijnenlaan 200d, 3001 Leuven, Belgium; IMEC, Kapeldreef 75, 3001 Leuven, Belgium.
| | | | | | | |
Collapse
|
6
|
Huang HP, Chiang W, Stone L, Kang CK, Chuang CY, Kuo HC. Using human Pompe disease-induced pluripotent stem cell-derived neural cells to identify compounds with therapeutic potential. Hum Mol Genet 2020; 28:3880-3894. [PMID: 31518394 DOI: 10.1093/hmg/ddz218] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/26/2019] [Accepted: 09/02/2019] [Indexed: 12/28/2022] Open
Abstract
Pompe disease (OMIM # 232300) is a glycogen storage disease caused by autosomal recessive mutations of the gene encoding alpha-1,4-glucosidase (GAA; EC 3.2.1.20). Despite the relatively effective employment of enzyme replacement therapy, some critical medical issues still exist in patients with this disease, including the persistence of abnormalities in the central nervous system (CNS), probably because of the inability of the recombinant GAA to pass through the blood-brain barrier. To address this issue, identification of more therapeutic agents that target the CNS of patients with Pompe disease may be required. In this study, we derived neuronal cells from Pompe disease-induced pluripotent stem cells (Pom-iPSCs) and proved that they are able to recapitulate the hallmark cellular and biochemical phenotypes of Pompe disease. Using the Pom-iPSC-derived neurons as an in vitro drug-testing model, we then identified three compounds, ebselen, wortmannin and PX-866, with therapeutic potential to alleviate Pompe disease-associated pathological phenotypes in the neurons derived from Pom-iPSCs. We confirmed that all three compounds were able to enhance the GAA activity in the Pom-iPSC-derived neurons. Moreover, they were able to enhance the GAA activity in several important internal organs of GAA-deficient mice when co-injected with recombinant human GAA, and we found that intraperitoneal injection of ebselen was able to promote the GAA activity of the GAA-heterozygous mouse brain. Our results prove the usefulness of Pom-iPSC-derived neuronal populations for identifying new compounds with therapeutic potential.
Collapse
Affiliation(s)
- Hsiang-Po Huang
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei, 10051, Taiwan
| | - Wei Chiang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Lee Stone
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Chun-Kai Kang
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei, 10051, Taiwan
| | - Ching-Yu Chuang
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Hung-Chih Kuo
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei, 10051, Taiwan.,Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan
| |
Collapse
|
7
|
Tabassum R, Jeong NY, Jung J. Therapeutic importance of hydrogen sulfide in age-associated neurodegenerative diseases. Neural Regen Res 2020; 15:653-662. [PMID: 31638087 PMCID: PMC6975154 DOI: 10.4103/1673-5374.266911] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 04/27/2019] [Accepted: 08/01/2019] [Indexed: 02/06/2023] Open
Abstract
Hydrogen sulfide (H2S) is a gasotransmitter that acts as an antioxidant and exhibits a wide variety of cytoprotective and physiological functions in age-associated diseases. One of the major causes of age-related diseases is oxidative stress. In recent years, the importance of H2S has become clear, although its antioxidant function has not yet been fully explored. The enzymes cystathionine β-synthase, cystathionine γ-lya-se, and 3-mercaptopyruvate sulfurtransferase are involved in the enzymatic production of H2S. Previously, H2S was considered a neuromodulator, given its role in long-term hippocampal potentiation, but it is now also recognized as an antioxidant in age-related neurodegeneration. Due to aerobic metabolism, the central nervous system is vulnerable to oxidative stress in brain aging, resulting in age-associated degenerative diseases. H2S exerts its antioxidant effect by limiting free radical reactions through the activation of antioxidant enzymes, including superoxide dismutase, catalase, and glutathione peroxidase, which protect against the effects of aging by regulating apoptosis-related genes, including p53, Bax, and Bcl-2. This review explores the implications and mechanisms of H2S as an antioxidant in age-associated neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, Huntington's disease, and Down syndrome.
Collapse
Affiliation(s)
- Rubaiya Tabassum
- Department of Anatomy and Cell Biology, College of Medicine, Dong-A University, Busan, Korea
- Department of Medicine, Graduate School, Dong-A University, Busan, Korea
| | - Na Young Jeong
- Department of Anatomy and Cell Biology, College of Medicine, Dong-A University, Busan, Korea
- Department of Medicine, Graduate School, Dong-A University, Busan, Korea
| | - Junyang Jung
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul, Korea
| |
Collapse
|
8
|
Abidar S, Yildiz O, Degirmenci A, Amakran A, El Maadoudi M, Nhiri M. Glucose-mediated protein glycation: Contribution of methanolic extract of Ceratonia siliqua L. in protection and in vitro potential inhibition of acetylcholinesterase. J Food Biochem 2019; 43:e13009. [PMID: 31393019 DOI: 10.1111/jfbc.13009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/02/2019] [Accepted: 07/12/2019] [Indexed: 01/11/2023]
Abstract
Chronic hyperglycemia presents the major etiology of diabetes mellitus and related complications mainly Alzheimer's disease, via the protein glycation and toxic products generated. In the current study, we investigated the eventual protective effect of the methanolic extract of Ceratonia siliqua L. (CsME) against glucose-mediated glycation in serum bovine albumin. The multi-stage glycation markers, namely fructosamines and advanced glycation end products (AGEs) levels were monitored along with measurement of thiol groups; moreover, the in vitro acetylcholinesterase (AChE) inhibition potential was carried out. HPLC was also assessed. Rutin was the main phenolic compound found in CsME. CsME showed a good capacity to inhibit AGEs, fructosamines and protected thiol groups against glycation. CsME exhibited a great AChE inhibition activity. In the present study, CsME prevented glucose-induced protein glycation, it also exhibited a good inhibition of AChE, suggesting its DM complications such as memory troubles related to AD. PRACTICAL APPLICATIONS: Neurodegenerative disorders ranging from memory troubles to Alzheimer's disease present the most diabetes mellitus complications and mainly attributed to protein glycation process. Currently, there is a strong trend to search for efficient natural sources of glycation and acetylcholinesterase inhibitors to replace the synthetic ones, whose secondary effects were shown. The present article tries to justify scientifically the wide use of Ceratonia siliqua L. in Moroccan folk medicine, demonstrating that the methanolic extract of leaves from this species presents a promising source of new natural compounds inhibiting acetylcholinesterase and acting in vitro against glycation generated compounds. Furthermore, for the first time, Rutin was the main phenolic compound found in this extract, these encouraging results should be coupled with further studies to integrate it in pharmaceutical formulations. As such, this paper should be of interest to a broad readership, including those interested in Biochemistry, Phytochemistry, pharmacology, and neurosciences.
Collapse
Affiliation(s)
- Sara Abidar
- Laboratoire de Biochimie et Génétique Moléculaire, Faculté des Sciences et Techniques, Université Abdelmalek Essaâdi, Tanger Principal, Morocco
| | - Oktay Yildiz
- Maçka VHS, Department of Food Processing, Karadeniz Technical University, Trabzon, Turkey
| | - Atiye Degirmenci
- Maçka VHS, Department of Food Processing, Karadeniz Technical University, Trabzon, Turkey
| | - Amina Amakran
- Laboratoire de Biochimie et Génétique Moléculaire, Faculté des Sciences et Techniques, Université Abdelmalek Essaâdi, Tanger Principal, Morocco
| | - Mohammed El Maadoudi
- Laboratoire Régional d'Analyses et de Recherches de l'ONSSA (office national de sécurité sanitaire des produits alimentaires), Tanger, Maroc
| | - Mohamed Nhiri
- Laboratoire de Biochimie et Génétique Moléculaire, Faculté des Sciences et Techniques, Université Abdelmalek Essaâdi, Tanger Principal, Morocco
| |
Collapse
|
9
|
Use of human pluripotent stem cell-derived cells for neurodegenerative disease modeling and drug screening platform. Future Med Chem 2019; 11:1305-1322. [DOI: 10.4155/fmc-2018-0520] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Most neurodegenerative diseases are characterized by a complex and mostly still unresolved pathology. This fact, together with the lack of reliable disease models, has precluded the development of effective therapies counteracting the disease progression. The advent of human pluripotent stem cells has revolutionized the field allowing the generation of disease-relevant neural cell types that can be used for disease modeling, drug screening and, possibly, cell transplantation purposes. In this Review, we discuss the applications of human pluripotent stem cells, the development of efficient protocols for the derivation of the different neural cells and their applicability for robust in vitro disease modeling and drug screening platforms for most common neurodegenerative conditions.
Collapse
|
10
|
Czerwińska P, Mazurek S, Kołodziejczak I, Wiznerowicz M. Gene delivery methods and genome editing of human pluripotent stem cells. Rep Pract Oncol Radiother 2019; 24:180-187. [PMID: 30820192 DOI: 10.1016/j.rpor.2019.01.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 11/21/2018] [Accepted: 01/27/2019] [Indexed: 12/24/2022] Open
Abstract
Induced pluripotent stem cells derived from normal somatic cells could be utilized to study tumorigenesis through overexpression of specific oncogenes, downregulation of tumor suppressors and dysregulation of other factors thought to promote tumorigenesis. Therefore, effective approaches that provide direct modifications of induced pluripotent stem cell genome are extremely needed. Emerging strategies are expected to provide the ability to more effectively introduce diverse genetic alterations, from as small as single-nucleotide modifications to whole gene amplification or deletion, all with a high degree of target specificity. To date, several techniques have been applied in stem cell studies to directly edit cell genome (ZFNs, TALENs or CRISPR/Cas9). In this review, we summarize specific gene delivery strategies that were applied to stem cell studies together with genome editing techniques, which enable a direct modification of endogenous DNA sequences in the context of cancer studies.
Collapse
Affiliation(s)
- Patrycja Czerwińska
- Laboratory of Gene Therapy, Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, Poznan, Poland.,Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, Poznan, Poland
| | - Sylwia Mazurek
- Laboratory of Gene Therapy, Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, Poznan, Poland.,Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, Poznan, Poland.,Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Iga Kołodziejczak
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, Poznan, Poland
| | - Maciej Wiznerowicz
- Laboratory of Gene Therapy, Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, Poznan, Poland.,Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
11
|
Yoshida K, Okada M, Nagasaka R, Sasaki H, Okada M, Kanie K, Kato R. Time-course colony tracking analysis for evaluating induced pluripotent stem cell culture processes. J Biosci Bioeng 2019; 128:209-217. [PMID: 30738731 DOI: 10.1016/j.jbiosc.2019.01.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/11/2019] [Accepted: 01/16/2019] [Indexed: 01/15/2023]
Abstract
Increasing the yield and maintaining a high quality of induced pluripotent stem cells (iPSCs) is necessary for manufacturing iPSCs at the industrial scale. However, because iPSCs are delicate, it is important to evaluate their quality during processing. To examine the status of cultured iPSCs non-invasively, morphology-based iPSC colony evaluation may be an efficient technology for cellular status monitoring and analysis. In this study, we examined the effectiveness of time-course colony tracking analysis for evaluating the iPSC culture process. Particularly, we obtained detailed time-course data to evaluate the effect of the pipetting technique on cell dissociation before seeding. Although the pipetting process causes severe shear stress to cells, which affects their quality, these effects have not been quantitatively analyzed because of their complex and uncontrollable parameters. By analyzing the heterogeneity and time-course responses of individual colonies, our colony tracking analysis revealed a critically damaged population caused by pipetting stress which could not be detected in conventional bulk analysis. Moreover, by comprehensively analyzing colony tracking data, which links the time-course morphology and marker staining results with each colony, we found that colony morphology is only highly correlated with the undifferentiated marker in the final stage, with a lower correlation in the early stages. Thus, colony tracking analysis provides a way to quantify cellular morphological information when evaluating complex iPSC manufacturing processes.
Collapse
Affiliation(s)
- Kei Yoshida
- Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Furocho, Chikusa-ku, Nagoya 464-8602, Japan
| | - Mika Okada
- Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Furocho, Chikusa-ku, Nagoya 464-8602, Japan
| | - Risako Nagasaka
- Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Furocho, Chikusa-ku, Nagoya 464-8602, Japan
| | - Hiroto Sasaki
- Department of Biotechnology, Graduate School of Engineering, Nagoya University, Furocho, Chikusa-ku, Nagoya 464-8602, Japan
| | - Mai Okada
- Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Furocho, Chikusa-ku, Nagoya 464-8602, Japan
| | - Kei Kanie
- Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Furocho, Chikusa-ku, Nagoya 464-8602, Japan
| | - Ryuji Kato
- Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Furocho, Chikusa-ku, Nagoya 464-8602, Japan; Stem Cell Evaluation Technology Research Association (SCA), Hacho-bori, Chuou-ku, Tokyo 104-0032, Japan; Institute of Nano-Life-Systems, Institute of Innovation for Future Society, Nagoya University, Division of Micro-Nano Mechatronics, Furocho, Chikusa-ku, Nagoya 464-8602, Japan.
| |
Collapse
|
12
|
Srinivasan G, Morgan D, Varun D, Brookhouser N, Brafman DA. An integrated biomanufacturing platform for the large-scale expansion and neuronal differentiation of human pluripotent stem cell-derived neural progenitor cells. Acta Biomater 2018; 74:168-179. [PMID: 29775730 DOI: 10.1016/j.actbio.2018.05.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 05/03/2018] [Accepted: 05/07/2018] [Indexed: 12/12/2022]
Abstract
Human pluripotent stem cell derived neural progenitor cells (hNPCs) have the unique properties of long-term in vitro expansion as well as differentiation into the various neurons and supporting cell types of the central nervous system (CNS). Because of these characteristics, hNPCs have tremendous potential in the modeling and treatment of various CNS diseases and disorders. However, expansion and neuronal differentiation of hNPCs in quantities necessary for these applications is not possible with current two dimensional (2-D) approaches. Here, we used a fully defined peptide substrate as the basis for a microcarrier (MC)-based suspension culture system. Several independently derived hNPC lines were cultured on MCs for multiple passages as well as efficiently differentiated to neurons. Finally, this MC-based system was used in conjunction with a low shear rotating wall vessel (RWV) bioreactor for the integrated, large-scale expansion and neuronal differentiation of hNPCs. Overall, this fully defined and scalable biomanufacturing system will facilitate the generation of hNPCs and their neuronal derivatives in quantities necessary for basic and translational applications. STATEMENT OF SIGNIFICANCE In this work, we developed a microcarrier (MC)-based culture system that allows for the expansion and neuronal differentiation of human pluripotent stem cell-derived neural progenitor cells (hNPCs) under defined conditions. In turn, this MC approach was implemented in a rotating wall vessel (RWV) bioreactor for the large-scale expansion and neuronal differentiation of hNPCs. This work is of significance as it overcomes current limitations of conventional two dimensional (2-D) culture systems to enable the generation of hNPCs and their neuronal derivatives in quantities required for downstream applications in disease modeling, drug screening, and regenerative medicine.
Collapse
Affiliation(s)
- Gayathri Srinivasan
- School of Biological and Health Systems Engineering, Arizona State University, United States
| | - Daylin Morgan
- School of Biological and Health Systems Engineering, Arizona State University, United States
| | - Divya Varun
- School of Biological and Health Systems Engineering, Arizona State University, United States
| | - Nicholas Brookhouser
- School of Biological and Health Systems Engineering, Arizona State University, United States
| | - David A Brafman
- School of Biological and Health Systems Engineering, Arizona State University, United States.
| |
Collapse
|
13
|
Affiliation(s)
- Anica Dricu
- a Department of Biochemistry , University of Medicine and Pharmacy of Craiova , Craiova , Romania
| |
Collapse
|
14
|
Yener Ilce B, Cagin U, Yilmazer A. Cellular reprogramming: A new way to understand aging mechanisms. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2018; 7. [PMID: 29350802 DOI: 10.1002/wdev.308] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 09/29/2017] [Accepted: 10/13/2017] [Indexed: 12/11/2022]
Abstract
Increased life expectancy, due to the rise in life quality and the decline in mortality rates, is leading to a society in which the population aged 60 and over is growing more rapidly than the entire population. Although various models and model organisms have been employed to investigate the mechanism of aging, induced pluripotent stem cells (iPSCs) are useful candidates to study human aging and age-related human diseases. This work discusses how iPSCs can be used as an alternative to the model organisms such as yeast, Caenorhabditis elegans, Drosophila melanogaster, or the mouse. The main focus is the reprogramming technology of somatic cells which is thought to provide an important perspective for rejuvenation strategies. The effects and relationships between aging and cell reprogramming are discussed, and studies related to aging and cell reprogramming are critically reviewed. We believe that for future studies, different parameters and detailed quantitative experiments should be performed in order to clearly understand the effect of aging on human cell reprogramming with respect to programming efficiency and differentiation capacity. This way, new insights will be provided to prevent or even reverse the aging process. WIREs Dev Biol 2018, 7:e308. doi: 10.1002/wdev.308 This article is categorized under: Adult Stem Cells, Tissue Renewal, and Regeneration > Stem Cells and Aging Adult Stem Cells, Tissue Renewal, and Regeneration > Regeneration Adult Stem Cells, Tissue Renewal, and Regeneration > Stem Cells and Disease.
Collapse
Affiliation(s)
| | | | - Acelya Yilmazer
- Biomedical Engineering Department, Engineering Faculty, Ankara University, Ankara, Turkey.,Stem Cell Institute, Ankara University, Ankara, Turkey
| |
Collapse
|
15
|
High-altitude adaptation in humans: from genomics to integrative physiology. J Mol Med (Berl) 2017; 95:1269-1282. [PMID: 28951950 DOI: 10.1007/s00109-017-1584-7] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 08/07/2017] [Accepted: 08/20/2017] [Indexed: 12/19/2022]
Abstract
About 1.2 to 33% of high-altitude populations suffer from Monge's disease or chronic mountain sickness (CMS). Number of factors such as age, sex, and population of origin (older, male, Andean) contribute to the percentage reported from a variety of samples. It is estimated that there are around 83 million people who live at altitudes > 2500 m worldwide and are at risk for CMS. In this review, we focus on a human "experiment in nature" in various high-altitude locations in the world-namely, Andean, Tibetan, and Ethiopian populations that have lived under chronic hypoxia conditions for thousands of years. We discuss the adaptive as well as mal-adaptive changes at the genomic and physiological levels. Although different genes seem to be involved in adaptation in the three populations, we can observe convergence at genetic and signaling, as well as physiological levels. What is important is that we and others have shown that lessons learned from the genes mined at high altitude can be helpful in better understanding and treating diseases that occur at sea level. We discuss two such examples: EDNRB and SENP1 and their role in cardiac tolerance and in the polycythemic response, respectively.
Collapse
|
16
|
Nagy J, Kobolák J, Berzsenyi S, Ábrahám Z, Avci HX, Bock I, Bekes Z, Hodoscsek B, Chandrasekaran A, Téglási A, Dezső P, Koványi B, Vörös ET, Fodor L, Szél T, Németh K, Balázs A, Dinnyés A, Lendvai B, Lévay G, Román V. Altered neurite morphology and cholinergic function of induced pluripotent stem cell-derived neurons from a patient with Kleefstra syndrome and autism. Transl Psychiatry 2017; 7:e1179. [PMID: 28742076 PMCID: PMC5538124 DOI: 10.1038/tp.2017.144] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 04/28/2017] [Indexed: 01/05/2023] Open
Abstract
The aim of the present study was to establish an in vitro Kleefstra syndrome (KS) disease model using the human induced pluripotent stem cell (hiPSC) technology. Previously, an autism spectrum disorder (ASD) patient with Kleefstra syndrome (KS-ASD) carrying a deleterious premature termination codon mutation in the EHMT1 gene was identified. Patient specific hiPSCs generated from peripheral blood mononuclear cells of the KS-ASD patient were differentiated into post-mitotic cortical neurons. Lower levels of EHMT1 mRNA as well as protein expression were confirmed in these cells. Morphological analysis on neuronal cells differentiated from the KS-ASD patient-derived hiPSC clones showed significantly shorter neurites and reduced arborization compared to cells generated from healthy controls. Moreover, density of dendritic protrusions of neuronal cells derived from KS-ASD hiPSCs was lower than that of control cells. Synaptic connections and spontaneous neuronal activity measured by live cell calcium imaging could be detected after 5 weeks of differentiation, when KS-ASD cells exhibited higher sensitivity of calcium responses to acetylcholine stimulation indicating a lower nicotinic cholinergic tone at baseline condition in KS-ASD cells. In addition, gene expression profiling of differentiated neuronal cells from the KS-ASD patient revealed higher expression of proliferation-related genes and lower mRNA levels of genes involved in neuronal maturation and migration. Our data demonstrate anomalous neuronal morphology, functional activity and gene expression in KS-ASD patient-specific hiPSC-derived neuronal cultures, which offers an in vitro system that contributes to a better understanding of KS and potentially other neurodevelopmental disorders including ASD.
Collapse
Affiliation(s)
- J Nagy
- Pharmacology and Drug Safety Research, Gedeon Richter Plc., Budapest, Hungary,Laboratory of Molecular Cell Biology, Gedeon Richter Plc. Gyömrői út 19-21., Budapest 1103, Hungary. E-mail:
| | | | - S Berzsenyi
- Pharmacology and Drug Safety Research, Gedeon Richter Plc., Budapest, Hungary
| | - Z Ábrahám
- Pharmacology and Drug Safety Research, Gedeon Richter Plc., Budapest, Hungary
| | - H X Avci
- BioTalentum Ltd., Gödöllő, Hungary
| | - I Bock
- BioTalentum Ltd., Gödöllő, Hungary
| | - Z Bekes
- Pharmacology and Drug Safety Research, Gedeon Richter Plc., Budapest, Hungary
| | - B Hodoscsek
- Pharmacology and Drug Safety Research, Gedeon Richter Plc., Budapest, Hungary
| | | | | | - P Dezső
- Pharmacology and Drug Safety Research, Gedeon Richter Plc., Budapest, Hungary
| | - B Koványi
- Pharmacology and Drug Safety Research, Gedeon Richter Plc., Budapest, Hungary
| | - E T Vörös
- Pharmacology and Drug Safety Research, Gedeon Richter Plc., Budapest, Hungary
| | - L Fodor
- Pharmacology and Drug Safety Research, Gedeon Richter Plc., Budapest, Hungary
| | - T Szél
- Pharmacology and Drug Safety Research, Gedeon Richter Plc., Budapest, Hungary
| | - K Németh
- Autism Foundation, Budapest, Hungary
| | - A Balázs
- Autism Foundation, Budapest, Hungary
| | | | - B Lendvai
- Pharmacology and Drug Safety Research, Gedeon Richter Plc., Budapest, Hungary
| | - G Lévay
- Pharmacology and Drug Safety Research, Gedeon Richter Plc., Budapest, Hungary
| | - V Román
- Pharmacology and Drug Safety Research, Gedeon Richter Plc., Budapest, Hungary
| |
Collapse
|
17
|
Suzuki S, Akamatsu W, Kisa F, Sone T, Ishikawa KI, Kuzumaki N, Katayama H, Miyawaki A, Hattori N, Okano H. Efficient induction of dopaminergic neuron differentiation from induced pluripotent stem cells reveals impaired mitophagy in PARK2 neurons. Biochem Biophys Res Commun 2017; 483:88-93. [PMID: 28057485 DOI: 10.1016/j.bbrc.2016.12.188] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 12/31/2016] [Indexed: 11/18/2022]
Abstract
Patient-specific induced pluripotent stem cells (iPSCs) show promise for use as tools for in vitro modeling of Parkinson's disease. We sought to improve the efficiency of dopaminergic (DA) neuron induction from iPSCs by the using surface markers expressed in DA progenitors to increase the significance of the phenotypic analysis. By sorting for a CD184high/CD44- fraction during neural differentiation, we obtained a population of cells that were enriched in DA neuron precursor cells and achieved higher differentiation efficiencies than those obtained through the same protocol without sorting. This high efficiency method of DA neuronal induction enabled reliable detection of reactive oxygen species (ROS) accumulation and vulnerable phenotypes in PARK2 iPSCs-derived DA neurons. We additionally established a quantitative system using the mt-mKeima reporter system to monitor mitophagy in which mitochondria fuse with lysosomes and, by combining this system with the method of DA neuronal induction described above, determined that mitophagy is impaired in PARK2 neurons. These findings suggest that the efficiency of DA neuron induction is important for the precise detection of cellular phenotypes in modeling Parkinson's disease.
Collapse
Affiliation(s)
- Sadafumi Suzuki
- Department of Physiology, Keio University, School of Medicine, Tokyo, Japan
| | - Wado Akamatsu
- Center for Genomic and Regenerative Medicine, Juntendo University, School of Medicine, Tokyo, Japan.
| | - Fumihiko Kisa
- Department of Physiology, Keio University, School of Medicine, Tokyo, Japan
| | - Takefumi Sone
- Department of Physiology, Keio University, School of Medicine, Tokyo, Japan
| | - Kei-Ichi Ishikawa
- Center for Genomic and Regenerative Medicine, Juntendo University, School of Medicine, Tokyo, Japan; Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Naoko Kuzumaki
- Department of Pharmacology, Hoshi University, Pharmacy and Pharmaceutical Sciences, Tokyo, Japan
| | - Hiroyuki Katayama
- Laboratory for Cell Function Dynamics, Brain Science Institute, RIKEN, Saitama, Japan
| | - Atsushi Miyawaki
- Laboratory for Cell Function Dynamics, Brain Science Institute, RIKEN, Saitama, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University, School of Medicine, Tokyo, Japan.
| |
Collapse
|
18
|
Zhang Q, Chen W, Tan S, Lin T. Stem Cells for Modeling and Therapy of Parkinson's Disease. Hum Gene Ther 2016; 28:85-98. [PMID: 27762639 DOI: 10.1089/hum.2016.116] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Parkinson's disease (PD) is the second most frequent neurodegenerative disease after Alzheimer's disease, which is characterized by a low level of dopamine being expressing in the striatum and a deterioration of dopaminergic neurons (DAn) in the substantia nigra pars compacta. Generation of PD-derived DAn, including differentiation of human embryonic stem cells, human neural stem cells, human-induced pluripotent stem cells, and direct reprogramming, provides an ideal tool to model PD, creating the possibility of mimicking key essential pathological processes and charactering single-cell changes in vitro. Furthermore, thanks to the understanding of molecular neuropathogenesis of PD and new advances in stem-cell technology, it is anticipated that optimal functionally transplanted DAn with targeted correction and transgene-free insertion will be generated for use in cell transplantation. This review elucidates stem-cell technology for modeling PD and offering desired safe cell resources for cell transplantation therapy.
Collapse
Affiliation(s)
- Qingxi Zhang
- 1 Center for Regenerative and Translational Medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine) , Guangzhou, China .,2 Department of Neurology, Zhujiang Hospital of Southern Medical University , Guangzhou, China
| | - Wanling Chen
- 1 Center for Regenerative and Translational Medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine) , Guangzhou, China .,2 Department of Neurology, Zhujiang Hospital of Southern Medical University , Guangzhou, China
| | - Sheng Tan
- 2 Department of Neurology, Zhujiang Hospital of Southern Medical University , Guangzhou, China
| | - Tongxiang Lin
- 1 Center for Regenerative and Translational Medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine) , Guangzhou, China .,3 Stem Cell Research Center, Fujian Agriculture and Forestry University , Fuzhou, China
| |
Collapse
|
19
|
Massa MG, Gisevius B, Hirschberg S, Hinz L, Schmidt M, Gold R, Prochnow N, Haghikia A. Multiple Sclerosis Patient-Specific Primary Neurons Differentiated from Urinary Renal Epithelial Cells via Induced Pluripotent Stem Cells. PLoS One 2016; 11:e0155274. [PMID: 27158987 PMCID: PMC4861271 DOI: 10.1371/journal.pone.0155274] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 04/26/2016] [Indexed: 12/11/2022] Open
Abstract
As multiple sclerosis research progresses, it is pertinent to continue to develop suitable paradigms to allow for ever more sophisticated investigations. Animal models of multiple sclerosis, despite their continuing contributions to the field, may not be the most prudent for every experiment. Indeed, such may be either insufficient to reflect the functional impact of human genetic variations or unsuitable for drug screenings. Thus, we have established a cell- and patient-specific paradigm to provide an in vitro model within which to perform future genetic investigations. Renal proximal tubule epithelial cells were isolated from multiple sclerosis patients’ urine and transfected with pluripotency-inducing episomal factors. Subsequent induced pluripotent stem cells were formed into embryoid bodies selective for ectodermal lineage, resulting in neural tube-like rosettes and eventually neural progenitor cells. Differentiation of these precursors into primary neurons was achieved through a regimen of neurotrophic and other factors. These patient-specific primary neurons displayed typical morphology and functionality, also staining positive for mature neuronal markers. The development of such a non-invasive procedure devoid of permanent genetic manipulation during the course of differentiation, in the context of multiple sclerosis, provides an avenue for studies with a greater cell- and human-specific focus, specifically in the context of genetic contributions to neurodegeneration and drug discovery.
Collapse
Affiliation(s)
- Megan G Massa
- Neurologische Klinik der Ruhr-Universität Bochum, St. Josef-Hospital, Bochum, Germany
| | - Barbara Gisevius
- Neurologische Klinik der Ruhr-Universität Bochum, St. Josef-Hospital, Bochum, Germany
| | - Sarah Hirschberg
- Neurologische Klinik der Ruhr-Universität Bochum, St. Josef-Hospital, Bochum, Germany
| | - Lisa Hinz
- Neurologische Klinik der Ruhr-Universität Bochum, St. Josef-Hospital, Bochum, Germany
| | - Matthias Schmidt
- Department of Neuroanatomy, Ruhr-Universität Bochum, Bochum, Germany
| | - Ralf Gold
- Neurologische Klinik der Ruhr-Universität Bochum, St. Josef-Hospital, Bochum, Germany
| | - Nora Prochnow
- Department of Neuroanatomy, Ruhr-Universität Bochum, Bochum, Germany
| | - Aiden Haghikia
- Neurologische Klinik der Ruhr-Universität Bochum, St. Josef-Hospital, Bochum, Germany
| |
Collapse
|
20
|
Csöbönyeiová M, Danišovič Ľ, Polák Š. Induced pluripotent stem cells for modeling and cell therapy of Parkinson's disease. Neural Regen Res 2016; 11:727-728. [PMID: 27335549 PMCID: PMC4904456 DOI: 10.4103/1673-5374.182692] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/12/2016] [Indexed: 12/22/2022] Open
Affiliation(s)
- Mária Csöbönyeiová
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Ľuboš Danišovič
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Štefan Polák
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| |
Collapse
|
21
|
Watson LM, Wong MMK, Becker EBE. Induced pluripotent stem cell technology for modelling and therapy of cerebellar ataxia. Open Biol 2016; 5:150056. [PMID: 26136256 PMCID: PMC4632502 DOI: 10.1098/rsob.150056] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Induced pluripotent stem cell (iPSC) technology has emerged as an important tool in understanding, and potentially reversing, disease pathology. This is particularly true in the case of neurodegenerative diseases, in which the affected cell types are not readily accessible for study. Since the first descriptions of iPSC-based disease modelling, considerable advances have been made in understanding the aetiology and progression of a diverse array of neurodegenerative conditions, including Parkinson's disease and Alzheimer's disease. To date, however, relatively few studies have succeeded in using iPSCs to model the neurodegeneration observed in cerebellar ataxia. Given the distinct neurodevelopmental phenotypes associated with certain types of ataxia, iPSC-based models are likely to provide significant insights, not only into disease progression, but also to the development of early-intervention therapies. In this review, we describe the existing iPSC-based disease models of this heterogeneous group of conditions and explore the challenges associated with generating cerebellar neurons from iPSCs, which have thus far hindered the expansion of this research.
Collapse
Affiliation(s)
- Lauren M Watson
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Maggie M K Wong
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Esther B E Becker
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
22
|
Deng W, Cao X, Wang Y, Yu Q, Zhang Z, Qu R, Chen J, Shao G, Gao X, Xu X, Yu J. Pleurotus eryngii Polysaccharide Promotes Pluripotent Reprogramming via Facilitating Epigenetic Modification. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2016; 64:1264-1273. [PMID: 26809505 DOI: 10.1021/acs.jafc.5b05661] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Pleurotus eryngii is a medicinal/edible mushroom with great nutritional value and bioactivity. Its polysaccharide has recently been developed into an effective gene vector via cationic modification. In the present study, cationized P. eryngii polysaccharide (CPS), hybridized with calcium phosphate (CP), was used to codeliver plasmids (Oct4, Sox2, Klf4, c-Myc) for generating induced pluripotent stem cells (iPSCs). The results revealed that the hybrid nanoparticles could significantly enhance the process and efficiency of reprogramming (1.6-fold increase) compared with the CP nanoparticles. The hybrid CPS also facilitated epigenetic modification during the reprogramming. Moreover, these hybrid nanoparticles exhibited multiple pathways (both caveolae- and clathrin-mediated endocytosis) in their cellular internalization, which accounted for the improved iPSCs generation. These findings therefore present a novel application of P. eryngii polysaccharide in pluripotent reprogramming via active epigenetic modification.
Collapse
Affiliation(s)
| | | | | | - Qingtong Yu
- School of Life Science & Technology, China Pharmaceutical University , Nanjing 210009, People's Republic of China
| | | | | | | | | | - Xiangdong Gao
- School of Life Science & Technology, China Pharmaceutical University , Nanjing 210009, People's Republic of China
| | | | | |
Collapse
|
23
|
Csöbönyeiová M, Polák Š, Danišovič L. Toxicity testing and drug screening using iPSC-derived hepatocytes, cardiomyocytes, and neural cells. Can J Physiol Pharmacol 2016; 94:687-94. [PMID: 27128322 DOI: 10.1139/cjpp-2015-0459] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Unexpected toxicity in areas such as cardiotoxicity, hepatotoxicity, and neurotoxicity is a serious complication of clinical therapy and one of the key causes for failure of promising drug candidates in development. Animal studies have been widely used for toxicology research to provide preclinical security evaluation of various therapeutic agents under development. Species differences in drug penetration of the blood-brain barrier, drug metabolism, and related toxicity contribute to failure of drug trials from animal models to human. The existing system for drug discovery has relied on immortalized cell lines, animal models of human disease, and clinical trials in humans. Moreover, drug candidates that are passed as being safe in the preclinical stage often show toxic effects during the clinical stage. Only around 16% drugs are approved for human use. Research on induced pluripotent stem cells (iPSCs) promises to enhance drug discovery and development by providing simple, reproducible, and economically effective tools for drug toxicity screening under development and, on the other hand, for studying the disease mechanism and pathways. In this review, we provide an overview of basic information about iPSCs, and discuss efforts aimed at the use of iPSC-derived hepatocytes, cardiomyocytes, and neural cells in drug discovery and toxicity testing.
Collapse
Affiliation(s)
- Mária Csöbönyeiová
- a Institute of Histology and Embryology, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 4, 811 08 Bratislava, Slovak Republic
| | - Štefan Polák
- a Institute of Histology and Embryology, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 4, 811 08 Bratislava, Slovak Republic
| | - L'uboš Danišovič
- b Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 4, 811 08 Bratislava, Slovak Republic
| |
Collapse
|
24
|
Telezhkin V, Schnell C, Yarova P, Yung S, Cope E, Hughes A, Thompson BA, Sanders P, Geater C, Hancock JM, Joy S, Badder L, Connor-Robson N, Comella A, Straccia M, Bombau G, Brown JT, Canals JM, Randall AD, Allen ND, Kemp PJ. Forced cell cycle exit and modulation of GABAA, CREB, and GSK3β signaling promote functional maturation of induced pluripotent stem cell-derived neurons. Am J Physiol Cell Physiol 2015; 310:C520-41. [PMID: 26718628 DOI: 10.1152/ajpcell.00166.2015] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 12/28/2015] [Indexed: 02/07/2023]
Abstract
Although numerous protocols have been developed for differentiation of neurons from a variety of pluripotent stem cells, most have concentrated on being able to specify effectively appropriate neuronal subtypes and few have been designed to enhance or accelerate functional maturity. Of those that have, most employ time courses of functional maturation that are rather protracted, and none have fully characterized all aspects of neuronal function, from spontaneous action potential generation through to postsynaptic receptor maturation. Here, we describe a simple protocol that employs the sequential addition of just two supplemented media that have been formulated to separate the two key phases of neural differentiation, the neurogenesis and synaptogenesis, each characterized by different signaling requirements. Employing these media, this new protocol synchronized neurogenesis and enhanced the rate of maturation of pluripotent stem cell-derived neural precursors. Neurons differentiated using this protocol exhibited large cell capacitance with relatively hyperpolarized resting membrane potentials; moreover, they exhibited augmented: 1) spontaneous electrical activity; 2) regenerative induced action potential train activity; 3) Na(+) current availability, and 4) synaptic currents. This was accomplished by rapid and uniform development of a mature, inhibitory GABAAreceptor phenotype that was demonstrated by Ca(2+) imaging and the ability of GABAAreceptor blockers to evoke seizurogenic network activity in multielectrode array recordings. Furthermore, since this protocol can exploit expanded and frozen prepatterned neural progenitors to deliver mature neurons within 21 days, it is both scalable and transferable to high-throughput platforms for the use in functional screens.
Collapse
Affiliation(s)
| | | | - Polina Yarova
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Sun Yung
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Emma Cope
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Alis Hughes
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | | | - Philip Sanders
- Department of Cell Biology, Immunology and Neuroscience, Faculty of Medicine, IDIBAPS, University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Charlene Geater
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Jane M Hancock
- School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; and
| | - Shona Joy
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Luned Badder
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | | | - Andrea Comella
- Department of Cell Biology, Immunology and Neuroscience, Faculty of Medicine, IDIBAPS, University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Marco Straccia
- Department of Cell Biology, Immunology and Neuroscience, Faculty of Medicine, IDIBAPS, University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Georgina Bombau
- Department of Cell Biology, Immunology and Neuroscience, Faculty of Medicine, IDIBAPS, University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Jon T Brown
- Hatherly Laboratory, Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, United Kingdom
| | - Josep M Canals
- Department of Cell Biology, Immunology and Neuroscience, Faculty of Medicine, IDIBAPS, University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Andrew D Randall
- School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; and Hatherly Laboratory, Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, United Kingdom
| | - Nicholas D Allen
- School of Biosciences, Cardiff University, Cardiff, United Kingdom;
| | - Paul J Kemp
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
25
|
Liu L, Huang JS, Han C, Zhang GX, Xu XY, Shen Y, Li J, Jiang HY, Lin ZC, Xiong N, Wang T. Induced Pluripotent Stem Cells in Huntington's Disease: Disease Modeling and the Potential for Cell-Based Therapy. Mol Neurobiol 2015; 53:6698-6708. [PMID: 26659595 DOI: 10.1007/s12035-015-9601-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 12/01/2015] [Indexed: 12/31/2022]
Abstract
Huntington's disease (HD) is an incurable neurodegenerative disorder that is characterized by motor dysfunction, cognitive impairment, and behavioral abnormalities. It is an autosomal dominant disorder caused by a CAG repeat expansion in the huntingtin gene, resulting in progressive neuronal loss predominately in the striatum and cortex. Despite the discovery of the causative gene in 1993, the exact mechanisms underlying HD pathogenesis have yet to be elucidated. Treatments that slow or halt the disease process are currently unavailable. Recent advances in induced pluripotent stem cell (iPSC) technologies have transformed our ability to study disease in human neural cells. Here, we firstly review the progress made to model HD in vitro using patient-derived iPSCs, which reveal unique insights into illuminating molecular mechanisms and provide a novel human cell-based platform for drug discovery. We then highlight the promises and challenges for pluripotent stem cells that might be used as a therapeutic source for cell replacement therapy of the lost neurons in HD brains.
Collapse
Affiliation(s)
- Ling Liu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jin-Sha Huang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chao Han
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Guo-Xin Zhang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiao-Yun Xu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yan Shen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jie Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hai-Yang Jiang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhi-Cheng Lin
- Department of Psychiatry, Harvard Medical School; Division of Alcohol and Drug Abuse, and Mailman Neuroscience Research Center, McLean Hospital, Belmont, MA, USA
| | - Nian Xiong
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tao Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
26
|
Wiethoff S, Arber C, Li A, Wray S, Houlden H, Patani R. Using human induced pluripotent stem cells to model cerebellar disease: hope and hype. J Neurogenet 2015; 29:95-102. [PMID: 25985846 PMCID: PMC4673530 DOI: 10.3109/01677063.2015.1053478] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 05/18/2015] [Indexed: 12/19/2022]
Abstract
The cerebellum forms a highly ordered and indispensible component of motor function within the adult neuraxis, consisting of several distinct cellular subtypes. Cerebellar disease, through a variety of genetic and acquired causes, results in the loss of function of defined subclasses of neurons, and remains a significant and untreatable health care burden. The scarcity of therapies in this arena can partially be explained by unresolved disease mechanisms due to inaccessibility of human cerebellar neurons in a relevant experimental context where initiating disease mechanisms could be functionally elucidated, or drug screens conducted. In this review we discuss the potential promise of human induced pluripotent stem cells (hiPSCs) for regenerative neurology, with a particular emphasis on in vitro modelling of cerebellar degeneration. We discuss progress made thus far using hiPSC-based models of neurodegeneration, noting the relatively slower pace of discovery made in modelling cerebellar dysfunction. We conclude by speculating how strategies attempting cerebellar differentiation from hiPSCs can be refined to allow the generation of accurate disease models. This in turn will permit a greater understanding of cerebellar pathophysiology to inform mechanistically rationalised therapies, which are desperately needed in this field.
Collapse
Affiliation(s)
- Sarah Wiethoff
- National Hospital for Neurology and Neurosurgery, UCL Institute of Neurology, London, UK
- Center for Neurology and Hertie Institute for Clinical Brain Research, Eberhard-Karls-University, Tübingen, Germany
| | - Charles Arber
- Department of Molecular Neuroscience and Queen Square Brain Bank, UCL Institute of Neurology, London, UK
| | - Abi Li
- Department of Molecular Neuroscience and Queen Square Brain Bank, UCL Institute of Neurology, London, UK
| | - Selina Wray
- Department of Molecular Neuroscience and Queen Square Brain Bank, UCL Institute of Neurology, London, UK
| | - Henry Houlden
- National Hospital for Neurology and Neurosurgery, UCL Institute of Neurology, London, UK
| | - Rickie Patani
- National Hospital for Neurology and Neurosurgery, UCL Institute of Neurology, London, UK
- Department of Molecular Neuroscience and Queen Square Brain Bank, UCL Institute of Neurology, London, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Euan MacDonald Centre for MND, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
27
|
Nityanandam A, Baldwin KK. Advances in reprogramming-based study of neurologic disorders. Stem Cells Dev 2015; 24:1265-83. [PMID: 25749371 DOI: 10.1089/scd.2015.0044] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The technology to convert adult human non-neural cells into neural lineages, through induced pluripotent stem cells (iPSCs), somatic cell nuclear transfer, and direct lineage reprogramming or transdifferentiation has progressed tremendously in recent years. Reprogramming-based approaches aimed at manipulating cellular identity have enormous potential for disease modeling, high-throughput drug screening, cell therapy, and personalized medicine. Human iPSC (hiPSC)-based cellular disease models have provided proof of principle evidence of the validity of this system. However, several challenges remain before patient-specific neurons produced by reprogramming can provide reliable insights into disease mechanisms or be efficiently applied to drug discovery and transplantation therapy. This review will first discuss limitations of currently available reprogramming-based methods in faithfully and reproducibly recapitulating disease pathology. Specifically, we will address issues such as culture heterogeneity, interline and inter-individual variability, and limitations of two-dimensional differentiation paradigms. Second, we will assess recent progress and the future prospects of reprogramming-based neurologic disease modeling. This includes three-dimensional disease modeling, advances in reprogramming technology, prescreening of hiPSCs and creating isogenic disease models using gene editing.
Collapse
Affiliation(s)
- Anjana Nityanandam
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, California
| | - Kristin K Baldwin
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, California
| |
Collapse
|
28
|
Young JE, Boulanger-Weill J, Williams DA, Woodruff G, Buen F, Revilla AC, Herrera C, Israel MA, Yuan SH, Edland SD, Goldstein LSB. Elucidating molecular phenotypes caused by the SORL1 Alzheimer's disease genetic risk factor using human induced pluripotent stem cells. Cell Stem Cell 2015; 16:373-85. [PMID: 25772071 DOI: 10.1016/j.stem.2015.02.004] [Citation(s) in RCA: 132] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 01/11/2015] [Accepted: 02/10/2015] [Indexed: 12/20/2022]
Abstract
Predisposition to sporadic Alzheimer's disease (SAD) involves interactions between a person's unique combination of genetic variants and the environment. The molecular effect of these variants may be subtle and difficult to analyze with standard in vitro or in vivo models. Here we used hIPSCs to examine genetic variation in the SORL1 gene and possible contributions to SAD-related phenotypes in human neurons. We found that human neurons carrying SORL1 variants associated with an increased SAD risk show a reduced response to treatment with BDNF, at the level of both SORL1 expression and APP processing. shRNA knockdown of SORL1 demonstrates that the differences in BDNF-induced APP processing between genotypes are dependent on SORL1 expression. We propose that the variation in SORL1 expression induction by BDNF is modulated by common genetic variants and can explain how genetic variation in this one locus can contribute to an individual's risk of developing SAD.
Collapse
Affiliation(s)
- Jessica E Young
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jonathan Boulanger-Weill
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Daniel A Williams
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Grace Woodruff
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Floyd Buen
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Arra C Revilla
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Cheryl Herrera
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Mason A Israel
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Shauna H Yuan
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Steven D Edland
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA; Division of Biostatistics, Department of Family and Preventive Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Lawrence S B Goldstein
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|